Structure-Based Virtual Screening Protocol for in Silico Identification of

Sep 26, 2016 - *Phone: +46-90-786-5266. ... A structure-based virtual screening (VS) protocol was developed with the intention of providing an efficie...
0 downloads 0 Views 1MB Size
Subscriber access provided by UNIV OF CALIFORNIA SAN DIEGO LIBRARIES

Article

A Structure-based Virtual Screening Protocol for in silico Identification of Potential Thyroid Disrupting Chemicals Targeting Transthyretin Jin Zhang, Afshan Begum, Kristoffer Brännström, Christin Grundström, Irina Iakovleva, Anders Olofsson, Astrid Elisabeth Sauer-Eriksson, and Patrik L. Andersson Environ. Sci. Technol., Just Accepted Manuscript • DOI: 10.1021/acs.est.6b02771 • Publication Date (Web): 26 Sep 2016 Downloaded from http://pubs.acs.org on September 28, 2016

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

Environmental Science & Technology is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 30

Environmental Science & Technology

1

A Structure-based Virtual Screening Protocol for in

2

silico Identification of Potential Thyroid Disrupting

3

Chemicals Targeting Transthyretin

4

Jin Zhang†, Afshan Begum†, Kristoffer Brännström‡, Christin Grundström†, Irina Iakovleva‡,

5

Anders Olofsson‡, A. Elisabeth Sauer-Eriksson†, Patrik L. Andersson†*

6

†Department of Chemistry, Umeå University, SE-901 87 Umeå, Sweden

7

‡Department of Medical Biochemistry and Biophysics, Umeå University, SE-901 87 Umeå,

8

Sweden

9

Corresponding Author

10

*Email: [email protected]. Phone: +46-90-786-5266, Fax: +46-90-786-7655

11

ACS Paragon Plus Environment

1

Environmental Science & Technology

Page 2 of 30

12

ABSTRACT

13

Thyroid disruption by xenobiotics is associated with a broad spectrum of severe adverse

14

outcomes. One possible molecular target of thyroid hormone disrupting chemicals (THDCs) is

15

transthyretin (TTR), a thyroid hormone transporter in vertebrates. To better understand the

16

interactions between TTR and THDCs, we determined the crystallographic structures of human

17

TTR in complex with perfluorooctanesulfonic acid (PFOS), perfluorooctanoic acid (PFOA), and

18

2,2',4,4'-tetrahydroxybenzophenone (BP2). The molecular interactions between the ligands and

19

TTR were further characterized using molecular dynamics simulations. A structure-based virtual

20

screening (VS) protocol was developed with the intention of providing an efficient tool for the

21

discovery of novel TTR-binders from the Tox21 inventory. Among the 192 predicted binders,

22

twelve representatives were selected and their TTR binding affinities were studied with

23

isothermal titration calorimetry, of which seven compounds had binding affinities between 0.26-

24

100 µM. To elucidate structural details in their binding to TTR, crystal structures were

25

determined of TTR in complex with four of the identified compounds including 2,6-dinitro-p-

26

cresol, bisphenol S, clonixin and triclopyr. The compounds were found to bind in the TTR

27

hormone binding sites as predicted. Our results show that the developed VS protocol is able to

28

successfully identify potential THDCs and we suggest that it can be used to propose THDCs for

29

future toxicological evaluations.

30

ACS Paragon Plus Environment

2

Page 3 of 30

Environmental Science & Technology

31

INTRODUCTION

32

Thyroid hormone disrupting chemicals (THDCs) are anthropogenic compounds that disrupt the

33

homeostasis of the thyroid hormone (TH) system1 and can induce disorders in physiological

34

processes, including macronutrient metabolism, energy balance, brain development, and

35

reproduction.2 Exposure to THDCs poses a significant threat to human health, especially during

36

fetal development. Prenatal hypothyroxinemia induced by THDCs can impair the development

37

of the embryonic brain and lead to neurologic deficits such as visuo-spatial processing

38

difficulties3 and reduced learning and memory.4

39

Transthyretin (TTR), a homotetrameric serum protein that transports L-thyroxine (T4),5 has

40

been suggested to be a possible molecular target of THDCs such as per and polyfluoroalkyl

41

substances (PFASs)6 and hydroxylated polychlorinated biphenyls (OH-PCBs).7 TTR is the

42

primary TH transporter in developing rodents,8 fish, birds, and amphibians9 and THDCs can

43

disrupt TH transport by competitively binding to the two identical thyroxine binding sites (TBSs)

44

situated at the dimeric interface of TTR.10,11 TTR can also mediate delivery of THDCs across the

45

placental and blood-brain barrier into the fetus and brain.12-14 Most TTR in human plasma resides

46

in the apo form and the concentration of tetrameric TTR in plasma (4-7 µM)15,16 is significantly

47

higher than that of T4 (0.112 µM)17. This means that low affinity TTR-binders can, without

48

competing with T4, bind to TTR and be transported to vital organs where they may subsequently

49

cause adverse health effects including developmental disorders.18,19 Inuit populations that are

50

chronically exposed to relatively high levels of TTR-binding contaminants showed normal T4

51

levels, but TTR-assisted accumulation of THDCs could potentially cause developmental

52

disorders.20-22 It is thus critical to understand the molecular interactions between THDCs and

53

TTR, and more importantly to identify and reduce the exposure to THDCs that target TTR.

ACS Paragon Plus Environment

3

Environmental Science & Technology

Page 4 of 30

54

Virtual screening (VS) using molecular docking is an efficient means to reduce costs and

55

animal testing by identifying the most hazardous compounds in risk assessment processes or

56

prioritizing the most promising candidates in drug discovery.23,24 The method has been used to

57

propose novel TTR amyloid inhibitors25 and to identify environmental pollutants targeting

58

estrogen receptor.26,27 Molecular docking can give insights into ligand-protein binding

59

conformations, and this was used to reveal interactions between PFASs and human liver fatty-

60

acid binding proteins.28 Such studies provide a better understanding of critical molecular

61

interactions between hazardous compounds and their targets.

62

Crystal structures of ligand-protein complexes are an essential component in the development

63

of VS protocols. Over 200 crystal structures of TTR have been reported in the Protein Data Bank

64

(PDB),29,30 but only a few of these are in complex with environmental pollutants or their

65

metabolites. These include TTR complexes with pentabromophenol31 and OH-PCBs.10 In

66

addition, we recently determined the crystal structure of TTR with the brominated flame

67

retardant tetrabromobisphenol A (TBBPA).32 Additional crystal structures of TTR complexes

68

with environmental pollutants with large chemical variation would improve the development of a

69

VS protocol for environmental pollutants and increase our understanding of their interactions

70

with TTR at the molecular level.

71

In this study, we determined the X-ray structures of human TTR in complex with three

72

emerging contaminants that were shown to bind to TTR: perfluorooctanoic acid (PFOA),

73

perfluorooctanesulfonic acid (PFOS), and 2,2’,4,4’-tetrahydroxybenzophenone (BP2).6,33 PFOA

74

and PFOS are commonly used as stain, water, and grease repellents in food packaging, carpets,

75

and textiles as well as in fire-fighting foams.34 BP2 is used as a UV absorber in personal care

76

products and plastic materials.35 Exposure to PFOS and PFOA is associated with hypothyroidism

ACS Paragon Plus Environment

4

Page 5 of 30

Environmental Science & Technology

77

and an increasing incidence of osteoporosis.36-38 BP2 might cause the development of

78

endometriosis.39 These ligands together with TBBPA cover a wide chemical variation of TTR

79

ligands. Their TTR binding activities in human plasma were measured with a previously

80

established plasma assay.32,40 The ligand-TTR interactions in the X-ray structures were further

81

studied by performing molecular dynamics (MD) simulations, and residues important for the

82

interactions were proposed by decomposition of ligand-binding free energies using the

83

Molecular Mechanics-Generalized Born Surface Area (MM-GBSA) method. Based on the novel

84

X-ray structures and the molecular interaction information, a structure-based VS protocol was

85

developed to identify THDCs targeting TTR from the Tox21 inventory of 7,849 organic

86

compounds.41 Twelve representative industrial compounds were selected for in vitro studies

87

using isothermal titration calorimetry (ITC). Of these, four hits have been co-crystallized with

88

TTR to study their binding conformations.

89

MATERIALS AND METHODS

90

Crystallization of the TTR-THDC complexes. Human wild-type TTR was purified and

91

crystallized in complex with PFOA, PFOS, BP2 and with four confirmed hits (bisphenol S

92

(BPS), clonixin, 2,6-dinitro-p-cresol (DNPC), and triclopyr) following the procedures described

93

previously.42,43 The purified TTR was dialyzed against 10 mM Na-phosphate buffer with 100

94

mM KCl (pH 7.6) and concentrated to 5 mg/mL using an Amicon Ultra centrifugal filter device

95

(Millipore, 3 kDa molecular-weight cutoff) and co-crystallized at room temperature with a 5-

96

molar excess of the compounds using the vapor-diffusion hanging drop method. A drop

97

containing 3 µL protein solution was mixed with 3 µL precipitant and equilibrated against 1 mL

98

reservoir solution containing 1.3–1.6 M sodium citrate and 3.5% v/v glycerol at pH 5.5 in 24-

ACS Paragon Plus Environment

5

Environmental Science & Technology

99 100

Page 6 of 30

well Linbro plates. Crystals grew to dimensions of 0.2 × 0.2 × 0.3 mm3 after 5 days. The crystals were cryoprotected with 12% v/v glycerol.

101

Crystallographic data collection, integration, and structure determination. The X-ray

102

diffraction data of the complexes were collected at the European Synchrotron Radiation Facility

103

(Grenoble, France) on beamline ID29 or ID23-1 and in-house instruments (Table S1 and S2).

104

The structure of human TTR (PDB ID: 1F415) and X-ray data from 38.2–2.5 Å resolution were

105

used in molecular replacement searches with the program PHASER.44 The models were refined

106

against all of the diffraction data using PHENIX.45 At the end of structure refinement,

107

anisotropic B-factors were refined. Manual map inspection was performed with COOT.46 The

108

details of the refinement statistics are shown in Table S1 and S2. Molecular graphics were

109

produced using CCP4mg.47 Structure factors and coordinates of the TTR complexes have been

110

deposited with the Protein Data Bank (PDB ID: 5JID (PFOA), 5JIM (PFOS), 5JIQ (BP2), 5LAF

111

(DNPC), 5L4I (clonixin), 5LAJ (BPS) and 5L4M (triclopyr)).

112

Molecular dynamics simulations. The MD simulations were prepared based on the three

113

structures of TTR complexes with BP2, PFOA (sharing similar binding profiles with PFOS), and

114

TBBPA32. Each system was prepared following the procedures described in the supporting

115

information. The MD simulations of each prepared system were performed using Amber 1448

116

with the following steps: 1) each system was minimized with two-stage energy minimizations, 2)

117

each system was gradually heated to 310 K and then equilibrated under NPT conditions for 1 ns,

118

and 3) the 20 ns production MD was performed under NPT conditions using a Langevin

119

thermostat and Berendsen barostat. Details of the simulation are described in the supporting

120

information.

ACS Paragon Plus Environment

6

Page 7 of 30

Environmental Science & Technology

121

MM-GBSA free-energy decomposition. We calculated the binding free energies of BP2,

122

PFOA, and TBBPA to TTR using the MM-GBSA method in AmberTools 1548 based on the last

123

10 ns of the MD trajectory. The contributions of each residue in the TBS to the ligand binding

124

were determined by the pairwise decomposition method. The five residues that contributed the

125

most to the enthalpic binding free energy of each ligand were considered to be important for

126

ligand-TTR interactions (Table S3). The entropy contributions were neglected due to low

127

prediction accuracy.49 The procedures and parameters are described in the supporting

128

information.

129

Collection and preparation of the ligand dataset. Two ligand sets were used in the study –

130

the TTR benchmarking set and the Tox21 inventory.41 The TTR benchmarking set (Table S4)

131

contained 155 TTR-binders and 10,197 in silico generated decoys (inactives).50 The TTR-binders

132

were collected from the scientific literature using an activity cutoff of Ki or IC50 ≤ 100 µM as

133

suggested by the US EPA in the studies of THDCs51 and estrogen disruptors26,52 and used in the

134

Tox21Challenge in silico model-developing exercise (more explanations given in supporting

135

information).53 The benchmarking set was used in the development and evaluation of the VS

136

protocol. The Tox21 dataset contains 7,849 industrial compounds and pharmaceuticals.54 For

137

each compound in the two sets, a maximum of 32 low-energy conformations were generated by

138

considering all chiral atoms using the Schrödinger LigPrep module under the OPLS_2005 force

139

field,55 and their ionization and tautomeric states were determined at pH 7.0 ± 1.0.

140

Molecular docking. Molecular docking models were developed using the TTR complex

141

structures with BP2, PFOA, and TBBPA. The TBS situated at the BB’ interface was described

142

by a grid box centered on its co-ligand. The prepared compounds in the TTR benchmarking set

143

were docked into each structure using the Schrödinger Glide module under standard precision.56

ACS Paragon Plus Environment

7

Environmental Science & Technology

Page 8 of 30

144

For the docking results, we assessed their VS performance and enrichment. The VS

145

performance of each model was described with the area under curve (AUC) value of the receiver

146

operating characteristic (ROC) curves. The AUC value was used because it is insensitive to the

147

ratio of actives versus decoys in the benchmarking set.57 The enrichment was characterized by

148

two enrichment factors (EFs) that were evaluated at 10% and 20% of the ranked database,

149

referred as EF10% and EF20%, respectively. The AUC values and EFs are given in Table S5.

150

Ligand-TTR interaction analysis. Residues important for ligand-TTR interactions were

151

identified using the MM-GBSA binding free-energy decomposition. The W188 water molecule

152

in the TTR-TBBPA complex was also considered because it was suggested to be critical for

153

mediating hydrogen bonds (H-bonds) between TBBPA and Ser117.32 For each initial hit

154

identified by molecular docking, we counted the number of electrostatic interactions (H-bonds

155

and salt bridges) with polar residues (including W188) and hydrophobic interactions with non-

156

polar residues based on their docking poses. The number of interactions together with the

157

docking score was used to refine the initial hits and identify the bioactive compounds from the

158

Tox21 inventory. The refined hits and their interactions with TTR are shown in Table S6. The

159

known TTR-binders identified among the refined hits are given in Table S7.

160

Measuring compound binding activity in human plasma. Binding activity of BP2, PFOS,

161

and PFOA to TTR in the presence of human plasma was measured with a previously established

162

plasma assay.40 Details of experimental procedures are given in supporting information.

163

Isothermal Titration Calorimetry. The binding affinities of twelve selected potential TTR-

164

binders were measured using an Auto-iTC200 (MicroCal, Malvern, UK) at 25 °C. The chemical

165

information and properties of the twelve compounds are shown in Table S8. Stock solutions

166

were prepared in dimethyl sulfoxide (DMSO) and diluted with phosphate-buffered saline (PBS,

ACS Paragon Plus Environment

8

Page 9 of 30

Environmental Science & Technology

167

pH=7.4) to a final level of 5% DMSO immediately before the ITC experiments.40 Aliquots of

168

each compound were titrated into the TTR buffer solution (30 or 90 µM) to reach a 10-fold molar

169

excess. For the control experiment, the compounds were titrated into the cell with only buffer.

170

Raw data were collected, subtracted for compound heats of dilution, and processed using the

171

MicroCal Origin software. Calorimetric data (Table S9) were plotted and fitted using the

172

standard single-site binding model to yield the binding affinities (Kd), enthalpy changes, and

173

entropy changes.

174

RESULTS AND DISCUSSION

175

Crystal structures of the TTR-THDC complexes. We co-crystallized human wild-type TTR

176

in complex with PFOA, PFOS, and BP2 and determined their structures at 1.2–1.4 Å resolution

177

(Table S1). The structures showed that all three ligands bind at the deep end of the TBS (Figure

178

1). Compared with T4, the three ligands have the ability to bind deeper in the pocket due to their

179

smaller molecular size (Figure S1).

180

Of particular interest are the TTR complex structures with PFOA and PFOS. The structures

181

share three unique features. 1) The two ligands bind with their hydrophobic tail inside the pocket

182

and their hydrophilic acid groups are surface exposed (Figure S2), which is different from our

183

previous assumption.33 2) The hydrophobic tail bends at the second to last carbon atom of the

184

ligand to accommodate the volume between two Ser117 residues and to allow the acid groups to

185

form salt bridges with Lys15. 3) The bent hydrophobic tail forces the hydroxyl groups on Ser117

186

to rotate away from the ligands (Figure 1), which creates a more hydrophobic pocket that favors

187

the ligand-TTR interactions.

188

BP2 interacts with TTR similarly as luteolin in the deep end of the TBS (Figure S2).58 Two

189

hydroxyl groups form hydrogen bonds with Ser117 residues, and the carbonyl group interacts

ACS Paragon Plus Environment

9

Environmental Science & Technology

Page 10 of 30

190

with Thr119. Interactions between the aromatic ring of BP2 and TTR involve van der Waal

191

contacts with the side chain of Leu17.

192

MD simulations for ligand-TTR interaction studies. We investigated the molecular

193

interactions between TTR and THDCs by performing MD simulations on the TTR complex

194

structures with BP2, PFOA, and TBBPA.32 All three ligands remained bound in the TBS, and the

195

root-mean-squared-deviation (RMSD) of the protein and ligands was under 2 Å throughout the

196

simulations (Figure S3 and Figure S4).

197

The ligand-TTR binding free energies were calculated using the MM-GBSA method based on

198

the last 10 ns of the MD trajectory. The calculated energies of the ligands correlate well with

199

their previously measured potencies (Figure S5).6,33,59 The energies were further decomposed to

200

reveal five residues of significance for the ligand-TTR interactions in each of the three complex

201

structures (Table S3). Two types of ligand-TTR interactions were identified electrostatic

202

interactions with the polar residues B-Thr119, B-Ser117, B’-Ser117, B-Lys15, and B’-Lys15 and

203

hydrophobic interactions with the non-polar residues B-Leu110, B-Leu17, B’-Leu17, and B-

204

Ala108. The electrostatic interactions account for specific ligand-TTR recognition and a large

205

proportion of the binding free energies,60-62 whereas the hydrophobic interactions were less

206

significant for ligand binding. The results also revealed that the three ligands interact with the

207

residues deeper in the TBS, e.g. Ser117, whereas T4 interacts with residues at the opening of the

208

TBS, e.g. Glu54.63 Information on the molecular interactions with the identified residues (Table

209

S3) was used to better understand the ligand-TTR interaction and to refine potential TTR-binders

210

for better identifying of bioactive compounds.

211

Molecular docking model. The accuracy of the molecular docking using Glide was assessed

212

by comparing the docking conformations of BP2, PFOA, and TBBPA with their X-ray

ACS Paragon Plus Environment

10

Page 11 of 30

Environmental Science & Technology

213

structures.33 The RMSD values for the co-ligands were lower than 2 Å (Table S5), indicating that

214

the molecular docking was able to reproduce the actual binding conformation. The prepared

215

compounds in the benchmarking set were docked into each TTR complex, and the docking

216

results were compared in terms of AUC values and enrichment factors (Table S5). The TTR-

217

TBBPA structure showed the best results with AUC of 0.75 and good early enrichment (EF10%

218

= 3.76 and EF20% = 2.95), and 141 binders were identified among the 155 binders in the

219

benchmark set (Table S5). The docking models based on the TTR-PFOA and TTR-BP2

220

structures had lower performances in identifying TTR-binders, probably because TBBPA best

221

reflects the majority of the halogenated and hydroxylated aromatic compounds in the benchmark

222

set. However, the two structures provide useful information for molecular interaction studies,

223

and the TTR-PFOA structure reveals the correct binding orientations of PFASs in the TBS where

224

the acidic group was found to interact with Lys15 rather than Ser117.

225

The impact of water in the TBS of the TTR-TBBPA complex was studied by evaluating the

226

model performance of the structure with and without water molecules in the TBS. The structure

227

with water in the TBS outperformed the one without (Table S5 and Figure S6). After examining

228

the ligand-protein interactions in the TTR-TBBPA structure, the W188 water molecule was

229

found to be critical for mediating hydrogen bonds between the ligand and the Ser117 residues,

230

which agrees well with previously reported data.32

231

Binding activity of the studied THDCs to TTR in human plasma. We compared the

232

binding activity of BP2, PFOA, and PFOS to TTR in human plasma in terms of their inhibitory

233

concentrations at 50% (IC50) to prevent tetrameric dissociation in the established assay.32,40 BP2

234

bound strongly to TTR with an IC50 of 4 µM, while PFOA (IC50 = 175 µM) and PFOS (IC50 =

235

103 µM) had poor binding activity to TTR in plasma (Figure S7). Their differences in binding

ACS Paragon Plus Environment

11

Environmental Science & Technology

Page 12 of 30

236

activity could be caused by the hydrophobic tails of the PFASs giving limited electrostatic

237

contributions to their TTR binding, which are critical for molecular recognition.40,62 TBBPA has

238

also shown strong binding to TTR (IC50 = 3.3 µM).32 Clearly, BP2 share characteristics with

239

TBBPA in terms of molecular interactions with TTR. It is thus of interest to identify

240

contaminants sharing similar molecular interactions with TTR as found in BP2 and TBBPA.

241

Virtual screening of industrial compounds. The docking model developed using the TTR-

242

TBBPA structure was used as the basis for the VS of the Tox21 inventory because of its high VS

243

performance and the strong binding of TBBPA to TTR in human plasma. We applied a two-step

244

procedure to identify potential TTR-binders from the Tox21 inventory. In the first step,

245

compounds were docked into the TTR-TBBPA structure followed by ranking based on their

246

standard precision docking score. For each industrial compound, all protonation states in the

247

range of pH 7.0±1.0 were considered during the molecular docking. As suggested by previous

248

studies,64,65 we selected its top scored protonation form (Table S4 and S6), since docking score

249

considers both binding affinity and tautomeric ratio.66,67 The top 20% scored compounds

250

covering 1,282 hits were considered as initial hits for TTR, and these were extracted for further

251

analysis. In the second step, the initial hits were further refined by selecting those that showed at

252

least three electrostatic interactions with the identified polar residues (including W188), and two

253

hydrophobic interactions with the non-polar residues. The criteria were based on the interactions

254

between TTR and TBBPA that binds to TTR with a Kd of 20 nM.32 The electrostatic interactions

255

have been suggested to be critical for specific recognition of TTR.60-62 A total of 192 substances

256

among the top 20% scored compounds also fulfilled the molecular interaction criteria (Table S6)

257

and thus were predicted to be potential TTR binders by the VS protocol. Eleven known TTR-

258

binders were identified (Table S7), including five pharmaceuticals (including diflunisal and

ACS Paragon Plus Environment

12

Page 13 of 30

Environmental Science & Technology

259

aceclofenac),40 two flame retardants (TBBPA and tetrachlorobisphenol A), one herbicide (2,4,5-

260

trichlorophenoxyacetic acid (2,4,5-T)),33 one UV filter BP2, and one T4 derivative (3,3´,5,5´-

261

tetraiodothyroacetic acid).68 The recognition of these known TTR binders by the VS protocol

262

indicates that it is capable of identifying potential TTR-binders for further toxicological

263

investigations.

264

Binding affinities of selected compounds. We selected twelve representative compounds

265

(Table 1) for in vitro validation from the VS results based on their various applications, structural

266

diversity, and environmental relevance. These compounds mainly represent herbicides and

267

polymer additives (plasticizers and polymerization inhibitors) and include primarily halogenated

268

and/or aromatic substances as well as nitro compounds. The thermodynamic profiles of the

269

ligand-TTR binding were investigated using ITC (Figure 2). One TTR tetramer can bind two

270

ligands. However, we fitted ITC thermograms (Figure 2) using a one-site binding model as

271

binding of the first ligand dominates the total binding energy and its affinity is approximately

272

100 times stronger than the second ligand.69,70 The ITC results showed that seven of the twelve

273

compounds bound to TTR with Kd values below 100 µM (Table 1) indicating that our VS

274

protocol gave a reasonable hit rate of approximately 60%, which can be compared with

275

commonly practiced VS campaigns that generally have a hit rate of 20-40%.71,72 The

276

thermodynamic data (Table S9) revealed that the binding of BPS, clonixin, fluroxypyr,

277

mesotrione, and triclopyr was largely driven by the enthalpic component, whereas the binding

278

energy of DNPC and picloram was mainly entropy driven. The affinities of the active

279

compounds were not correlated with their docking scores (Figure S8), which was not surprising

280

since docking scores generally give poor estimations of the binding affinities.73,74 However, the

281

trend is correct (except DNPC), as more negative docking scores are associated with higher

ACS Paragon Plus Environment

13

Environmental Science & Technology

Page 14 of 30

282

affinities. Notably, five predicted binders showed no activities in the ITC experiments (Figure

283

S9) and we provide a thorough discussion on the potential reasons for their mispredictions in the

284

supporting information.

285

Crystal structures of the TTR-confirmed hits complexes. Four confirmed TTR-binders

286

(clonixin, DNPC, triclopyr and BPS) were selected for co-crystallization with human wild-type

287

TTR, considering their high binding affinities, diverse structures and various applications. The

288

crystal structures were determined at 1.5-1.6 Å resolution (Table S2). Compared with T4, the

289

four ligands bind deeper in the TBS due to their smaller molecular size (Figure 3). Clonixin

290

interacts with TTR similarly as flufenamic acid.75 It forms H-bonds and salt bridges with Lys15

291

(Figure S10). DNPC presents different binding conformations in the two TBSs. Furthermore, in

292

the AA’ interface, two DNPC molecules bind into the TBS simultaneously where one DNPC

293

interacts with Ser117 at the deep end of TBS, whereas the other DNPC binds to Lys15 at the

294

cavity entrance (Figure S10). In the BB’ interface, DNPC also binds in two different

295

orientations, however these conformations are different from the ones in AA’ and both cannot be

296

occupied at the same time as their binding sites overlap (Figure 3). The multiple conformations

297

of DNPC give better knowledge on interactions between TTR and nitro compounds, which

298

provides guidance for molecular design and risk assessment. Triclopyr interacts with TTR by

299

having electrostatic interactions with Lys15. BPS binds in the TBS similarly as TBBPA.32 Its

300

hydroxyl groups interact with Lys15 and form water-bridged H-bonds with Thr119 and Ser117

301

(Figure S8). We compared the docking pose of each ligand with its crystal conformation in the

302

BB’ interface (Figure S11) and the results showed that we correctly predicted the binding

303

conformations for clonixin, DNPC, and triclopyr. The inaccurate prediction of BPS may be due

304

to its weaker affinity and high mobility in the TBS. Besides DNPC, EMD21388 was also

ACS Paragon Plus Environment

14

Page 15 of 30

Environmental Science & Technology

305

reported to adapt to distinctive binding-modes in the two TBSs.76 Ensemble docking has been

306

proposed as a suitable strategy for predicting conformation of such ligands,77,78 which however

307

was not completed here due to lack of TTR structures with similar binding-modes to DNPC.

308

Environmental implications of the potential THDCs. Seven compounds with diverse

309

structures and various applications were identified as TTR-binders (Table 1). These compounds

310

warrant further toxicological investigations considering the following two reasons: (1) humans

311

may be chronically exposed to the compounds at doses that are similar or significantly higher

312

than normal T4 level (0.112 µM)17. In vivo levels of the compounds have been reported to be

313

35844 µM (9.416 mg/ml) for clonixin79, 0.312 µM (0.08 µg/ml) for triclopyr80, 3.72 µM (0.93

314

µg/ml) for BPS81, and 9.19 µM (2.22 µg/ml) for picloram82. (2) Low-affinity binders could be

315

effectively transported by TTR and accumulated in vulnerable organs in vivo,12-14 since most

316

TTR in human plasma resides in the apo form.15-17 Their accumulations could subsequently

317

induce adverse effects including developmental disorders.18,19

318

Interestingly, BPS, a replacement for bisphenol A (BPA), bound to TTR with a Kd of 52 µM.

319

BPS is used extensively as a plasticizer in BPA-free consumer products and as an anticorrosive

320

agent in canned food. Despite claims that it is a safer alternative to BPA, BPS has been reported

321

to pose similar potential health hazards as BPA in a number of in vitro and in vivo studies.83

322

Embryonic exposure to BPS is associated with neurogenesis within the hypothalamus, causing

323

gestation and hyperactivity in juvenile zebrafish84 and non-associative learning impairments in

324

adult zebrafish.85 BPS can also decrease plasma TH levels and impair the reproductive potential

325

of zebrafish.86 It was also found in dust samples in twelve countries, and the highest median

326

concentration was in Greece (860 ng/g).87

ACS Paragon Plus Environment

15

Environmental Science & Technology

Page 16 of 30

327

Clonixin was the strongest TTR binder identified in the Tox21 inventory and had a Kd of 0.26

328

µM. Clonixin is a non-steroidal anti-inflammatory drug, and it has a similar structure and TTR

329

binding affinity as meclofenamic acid.40 To our knowledge, its thyroid effects have not been

330

studied.

331

DNPC is used as an inhibitor of styrene polymerization and a dye intermediate, and this

332

molecule showed the second strongest interaction with TTR. DNPC is an uncoupler of oxidative

333

phosphorylation in humans, and it can cause acute hyperthermia, kidney and liver failure, and

334

cerebral edema.88 DNPC has a high potential for accumulation in biota and humans.88

335

The herbicides fluroxypyr, mesotrione, picloram, and triclopyr were identified as TTR-binders,

336

with triclopyr being the strongest binder. Triclopyr is used as an alternative to 2,4,5-T, and it has

337

been reported to cause thyroid-related neurotoxicity89 and developmental toxicity in rats.90 The

338

major metabolite of triclopyr is 3,5,6-trichloro-2-pyridinol,91 which is also a TTR-binder,33 and it

339

has been reported to disrupt the levels of T4 and thyroid stimulating hormone in humans.92

340

Exposure to triclopyr poses a significant threat to its applicators, and the amount in their urine

341

samples was reported to be 1–13 mg/day.93 The compound has been detected in water samples

342

from Australia,94 France,95 and multiple locations in the US.96,97 Environmental implications of

343

the other three herbicides are given in the supporting information.

344

In summary, we have determined the binding conformations of PFOS, PFOA, and BP2 in the

345

TBS of TTR. The structures of the complexes provide valuable information for improving our

346

understanding of the molecular interactions between TTR and emerging contaminants. PFOS and

347

PFOA were previously reported as strong TTR-binders, but our results indicate that they are

348

weak binders in plasma, whereas BP2 showed strong binding activity . BP2 is thus more likely to

349

be transported by TTR and accumulated in vulnerable organs. Based on the novel TTR complex

ACS Paragon Plus Environment

16

Page 17 of 30

Environmental Science & Technology

350

structures and the information on molecular interactions obtained from MD simulations, a VS

351

protocol was developed for the identification of potential THDCs targeting TTR from the Tox21

352

inventory. The protocol proposed 192 industrial compounds as potential binders to TTR. Among

353

the twelve compounds tested using in vitro experiments, we identified seven novel TTR-binders

354

including clonixin, DNPC, triclopyr, fluroxypyr, BPS, picloram, and mesotrione. Most of these

355

have been detected in the environment and have shown adverse thyroid-related effects in animal

356

models. We further co-crystallized TTR with PBS, clonixin, DNPC, and triclopyr and their

357

structures showed that the compounds bind in the TBS as predicted by the VS protocol. We

358

suggest further in vivo studies on these TTR-targeting compounds especially as they may

359

accumulate in vulnerable organs.

360

Supporting Information

361

Details of the X-ray structures, plasma assay, and VS performance; the identified potential TTR-

362

binders and the compounds tested using ITC; procedures and results of the MD simulations and

363

MM-GBSA calculations can be found in the supporting information. This information is

364

available free of charge via the Internet at http://pubs.acs.org/.

365

ACKNOWLEDGMENT

366

This study was financed by the MiSSE project through grants from the Swedish Research

367

Council for the Environment, Agricultural Sciences and Spatial Planning (Formas) (210-2012-

368

131) and by the Swedish Research Council (VR) (521-2011-6427). The MD simulations were

369

conducted using the High Performance Computing Center North.

370

ACS Paragon Plus Environment

17

Environmental Science & Technology

Page 18 of 30

371 372

REFERENCES

373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412

(1) Murk, A. J.; Rijntjes, E.; Blaauboer, B. J.; Clewell, R.; Crofton, K. M.; Dingemans, M. M.; Furlow, J. D.; Kavlock, R.; Kohrle, J.; Opitz, R.; Traas, T.; Visser, T. J.; Xia, M.; Gutleb, A. C., Mechanism-based testing strategy using in vitro approaches for identification of thyroid hormone disrupting chemicals. Toxicol. In Vitro 2013, 27, (4), 1320-46. (2) Crofton, K. M., Thyroid disrupting chemicals: mechanisms and mixtures. Int. J. Androl. 2008, 31, (2), 209-23. (3) Haddow, J. E.; Palomaki, G. E.; Allan, W. C.; Williams, J. R.; Knight, G. J.; Gagnon, J.; O'Heir, C. E.; Mitchell, M. L.; Hermos, R. J.; Waisbren, S. E.; Faix, J. D.; Klein, R. Z., Maternal thyroid deficiency during pregnancy and subsequent neuropsychological development of the child. N. Engl. J. Med. 1999, 341, (8), 549-55. (4) Gilbert, M. E.; Rovet, J.; Chen, Z.; Koibuchi, N., Developmental thyroid hormone disruption: prevalence, environmental contaminants and neurodevelopmental consequences. Neurotoxicology 2012, 33, (4), 842-52. (5) Hornberg, A.; Eneqvist, T.; Olofsson, A.; Lundgren, E.; Sauer-Eriksson, A. E., A comparative analysis of 23 structures of the amyloidogenic protein transthyretin. J. Mol. Biol. 2000, 302, (3), 649-69. (6) Weiss, J. M.; Andersson, P. L.; Lamoree, M. H.; Leonards, P. E.; van Leeuwen, S. P.; Hamers, T., Competitive binding of poly- and perfluorinated compounds to the thyroid hormone transport protein transthyretin. Toxicol. Sci. 2009, 109, (2), 206-16. (7) Cheek, A. O.; Kow, K.; Chen, J.; McLachlan, J. A., Potential mechanisms of thyroid disruption in humans: interaction of organochlorine compounds with thyroid receptor, transthyretin, and thyroid-binding globulin. Environ. Health Perspect. 1999, 107, (4), 273-8. (8) Lans, M. C.; Spiertz, C.; Brouwer, A.; Koeman, J. H., Different competition of thyroxine binding to transthyretin and thyroxine-binding globulin by hydroxy-PCBs, PCDDs and PCDFs. Eur. J. Pharmacol. 1994, 270, (2-3), 129-36. (9) Schreiber, G., The evolutionary and integrative roles of transthyretin in thyroid hormone homeostasis. J. Endocrinol. 2002, 175, (1), 61-73. (10) Purkey, H. E.; Palaninathan, S. K.; Kent, K. C.; Smith, C.; Safe, S. H.; Sacchettini, J. C.; Kelly, J. W., Hydroxylated polychlorinated biphenyls selectively bind transthyretin in blood and inhibit amyloidogenesis: rationalizing rodent PCB toxicity. Chem. Biol. 2004, 11, (12), 1719-28. (11) Eneqvist, T.; Lundberg, E.; Karlsson, A.; Huang, S.; Santos, C. R.; Power, D. M.; SauerEriksson, A. E., High resolution crystal structures of piscine transthyretin reveal different binding modes for triiodothyronine and thyroxine. J. Biol. Chem. 2004, 279, (25), 26411-6. (12) Kim, S. Y.; Choi, E. S.; Lee, H. J.; Moon, C.; Kim, E., Transthyretin as a new transporter of nanoparticles for receptor-mediated transcytosis in rat brain microvessels. Colloids Surf. B. Biointerfaces 2015, 136, 989-96. (13) Meerts, I. A.; Assink, Y.; Cenijn, P. H.; Van Den Berg, J. H.; Weijers, B. M.; Bergman, A.; Koeman, J. H.; Brouwer, A., Placental transfer of a hydroxylated polychlorinated biphenyl and effects on fetal and maternal thyroid hormone homeostasis in the rat. Toxicol. Sci. 2002, 68, (2), 361-71.

ACS Paragon Plus Environment

18

Page 19 of 30

413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458

Environmental Science & Technology

(14) Diamanti-Kandarakis, E.; Bourguignon, J. P.; Giudice, L. C.; Hauser, R.; Prins, G. S.; Soto, A. M.; Zoeller, R. T.; Gore, A. C., Endocrine-disrupting chemicals: an Endocrine Society scientific statement. Endocr. Rev. 2009, 30, (4), 293-342. (15) Ritchie, R. F.; Palomaki, G. E.; Neveux, L. M.; Navolotskaia, O.; Ledue, T. B.; Craig, W. Y., Reference distributions for the negative acute-phase serum proteins, albumin, transferrin and transthyretin: a practical, simple and clinically relevant approach in a large cohort. J. Clin. Lab. Anal. 1999, 13, (6), 273-9. (16) Ritchie, R. F.; Palomaki, G. E.; Neveux, L. M.; Navolotskaia, O., Reference distributions for the negative acute-phase proteins, albumin, transferrin, and transthyretin: a comparison of a large cohort to the world's literature. J. Clin. Lab. Anal. 1999, 13, (6), 280-6. (17) Hollowell, J. G.; Staehling, N. W.; Flanders, W. D.; Hannon, W. H.; Gunter, E. W.; Spencer, C. A.; Braverman, L. E., Serum TSH, T(4), and thyroid antibodies in the United States population (1988 to 1994): National Health and Nutrition Examination Survey (NHANES III). J. Clin. Endocrinol. Metab. 2002, 87, (2), 489-99. (18) Boas, M.; Feldt-Rasmussen, U.; Main, K. M., Thyroid effects of endocrine disrupting chemicals. Mol. Cell. Endocrinol. 2012, 355, (2), 240-248. (19) Porterfield, S. P., Thyroidal dysfunction and environmental chemicals--potential impact on brain development. Environ. Health Perspect. 2000, 108 Suppl 3, 433-8. (20) Dallaire, R.; Muckle, G.; Dewailly, E.; Jacobson, S. W.; Jacobson, J. L.; Sandanger, T. M.; Sandau, C. D.; Ayotte, P., Thyroid hormone levels of pregnant inuit women and their infants exposed to environmental contaminants. Environ. Health Perspect. 2009, 117, (6), 1014-20. (21) Audet-Delage, Y.; Ouellet, N.; Dallaire, R.; Dewailly, E.; Ayotte, P., Persistent organic pollutants and transthyretin-bound thyroxin in plasma of Inuit women of childbearing age. Environ. Sci. Technol. 2013, 47, (22), 13086-92. (22) Plusquellec, P.; Muckle, G.; Dewailly, E.; Ayotte, P.; Begin, G.; Desrosiers, C.; Despres, C.; Saint-Amour, D.; Poitras, K., The relation of environmental contaminants exposure to behavioral indicators in Inuit preschoolers in Arctic Quebec. Neurotoxicology 2010, 31, (1), 17-25. (23) Rabinowitz, J. R.; Goldsmith, M. R.; Little, S. B.; Pasquinelli, M. A., Computational molecular modeling for evaluating the toxicity of environmental chemicals: prioritizing bioassay requirements. Environ. Health Perspect. 2008, 116, (5), 573-7. (24) Muralidharan, A. R.; Selvaraj, C.; Singh, S. K.; Sheu, J. R.; Thomas, P. A.; Geraldine, P., Structure-Based Virtual Screening and Biological Evaluation of a Calpain Inhibitor for Prevention of Selenite-Induced Cataractogenesis in an in Vitro System. J. Chem. Inf. Model. 2015, 55, (8), 1686-97. (25) Simoes, C. J.; Mukherjee, T.; Brito, R. M.; Jackson, R. M., Toward the discovery of functional transthyretin amyloid inhibitors: application of virtual screening methods. J. Chem. Inf. Model. 2010, 50, (10), 1806-20. (26) Rabinowitz, J. R.; Little, S. B.; Laws, S. C.; Goldsmith, M. R., Molecular modeling for screening environmental chemicals for estrogenicity: use of the toxicant-target approach. Chem. Res. Toxicol. 2009, 22, (9), 1594-602. (27) McRobb, F. M.; Kufareva, I.; Abagyan, R., In silico identification and pharmacological evaluation of novel endocrine disrupting chemicals that act via the ligand-binding domain of the estrogen receptor alpha. Toxicol. Sci. 2014, 141, (1), 188-97. (28) Zhang, L.; Ren, X. M.; Guo, L. H., Structure-based investigation on the interaction of perfluorinated compounds with human liver fatty acid binding protein. Environ. Sci. Technol. 2013, 47, (19), 11293-301.

ACS Paragon Plus Environment

19

Environmental Science & Technology

459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503

Page 20 of 30

(29) RCSB-PDB, RCSB Protein Data Bank (PDB). http://www.rcsb.org/pdb/ 2015. (30) Palaninathan, S. K., Nearly 200 X-ray crystal structures of transthyretin: what do they tell us about this protein and the design of drugs for TTR amyloidoses? Curr. Med. Chem. 2012, 19, (15), 2324-42. (31) Ghosh, M.; Meerts, I. A.; Cook, A.; Bergman, A.; Brouwer, A.; Johnson, L. N., Structure of human transthyretin complexed with bromophenols: a new mode of binding. Acta Crystallogr. D Biol. Crystallogr. 2000, 56, (Pt 9), 1085-95. (32) Iakovleva, I.; Begum, A.; Brannstrom, K.; Wijsekera, A.; Nilsson, L.; Zhang, J.; Andersson, P. L.; Sauer-Eriksson, A. E.; Olofsson, A., Tetrabromobisphenol A Is an Efficient Stabilizer of the Transthyretin Tetramer. PLoS One 2016, 11, (4), e0153529. (33) Zhang, J.; Kamstra, J. H.; Ghorbanzadeh, M.; Weiss, J. M.; Hamers, T.; Andersson, P. L., In Silico Approach To Identify Potential Thyroid Hormone Disruptors among Currently Known Dust Contaminants and Their Metabolites. Environ. Sci. Technol. 2015, 49, (16), 10099-107. (34) US-EPA Emerging Contaminants-Perfluorooctane Sulfonate (PFOS)and Perfluorooctanoic Acid (PFOA); The United States Environmental Protection Agency, 2014. (35) Spyropoulos, D. V., Stability testing of the plastics additives 2,4-dihydroxybenzophenone, 2,2 '-dihydroxy-4-methoxybenzophenone, 2-hydroxy-4-n-octyloxybenzophenone, 2-ethoxy-2ethyloxanilide (tinnvin 312) and 2,2 '-methylenebis(4-methyl-t-tert-butyl phenol) in aqueous and fatty food simulants. Food Addit. Contam. 1998, 15, (3), 362-369. (36) Melzer, D.; Rice, N.; Depledge, M. H.; Henley, W. E.; Galloway, T. S., Association between serum perfluorooctanoic acid (PFOA) and thyroid disease in the U.S. National Health and Nutrition Examination Survey. Environ. Health Perspect. 2010, 118, (5), 686-92. (37) Lopez-Espinosa, M. J.; Mondal, D.; Armstrong, B.; Bloom, M. S.; Fletcher, T., Thyroid function and perfluoroalkyl acids in children living near a chemical plant. Environ. Health Perspect. 2012, 120, (7), 1036-41. (38) Khalil, N.; Chen, A.; Lee, M.; Czerwinski, S. A.; Ebert, J. R.; DeWitt, J. C.; Kannan, K., Association of Perfluoroalkyl Substances, Bone Mineral Density, and Osteoporosis in the U.S. Population in NHANES 2009-2010. Environ. Health Perspect. 2016, 124, (1), 81-7. (39) Kunisue, T.; Chen, Z.; Buck Louis, G. M.; Sundaram, R.; Hediger, M. L.; Sun, L.; Kannan, K., Urinary concentrations of benzophenone-type UV filters in U.S. women and their association with endometriosis. Environ. Sci. Technol. 2012, 46, (8), 4624-32. (40) Iakovleva, I.; Brannstrom, K.; Nilsson, L.; Gharibyan, A. L.; Begum, A.; Anan, I.; Walfridsson, M.; Sauer-Eriksson, A. E.; Olofsson, A., Enthalpic Forces Correlate with the Selectivity of Transthyretin-Stabilizing Ligands in Human Plasma. J. Med. Chem. 2015, 58, (16), 6507-15. (41) US-NRC Toxicity Testing in the 21st Century: a Vision and a Strategy.; National Research Council. : National Academies Press. Washington, DC, 2007. (42) Johnson, S. M.; Connelly, S.; Wilson, I. A.; Kelly, J. W., Toward optimization of the second aryl substructure common to transthyretin amyloidogenesis inhibitors using biochemical and structural studies. J. Med. Chem. 2009, 52, (4), 1115-25. (43) Lindhagen-Persson, M.; Vestling, M.; Reixach, N.; Olofsson, A., Formation of cytotoxic transthyretin is not dependent on inter-molecular disulphide bridges commonly found within the amyloid form. Amyloid 2008, 15, (4), 240-5. (44) McCoy, A. J.; Grosse-Kunstleve, R. W.; Adams, P. D.; Winn, M. D.; Storoni, L. C.; Read, R. J., Phaser crystallographic software. J. Appl. Crystallogr. 2007, 40, (Pt 4), 658-674.

ACS Paragon Plus Environment

20

Page 21 of 30

504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547

Environmental Science & Technology

(45) Adams, P. D.; Afonine, P. V.; Bunkoczi, G.; Chen, V. B.; Davis, I. W.; Echols, N.; Headd, J. J.; Hung, L. W.; Kapral, G. J.; Grosse-Kunstleve, R. W.; McCoy, A. J.; Moriarty, N. W.; Oeffner, R.; Read, R. J.; Richardson, D. C.; Richardson, J. S.; Terwilliger, T. C.; Zwart, P. H., PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr. D Biol. Crystallogr. 2010, 66, (Pt 2), 213-21. (46) Emsley, P.; Cowtan, K., Coot: model-building tools for molecular graphics. Acta Crystallogr. D Biol. Crystallogr. 2004, 60, (Pt 12 Pt 1), 2126-32. (47) McNicholas, S.; Potterton, E.; Wilson, K. S.; Noble, M. E., Presenting your structures: the CCP4mg molecular-graphics software. Acta Crystallogr. D Biol. Crystallogr. 2011, 67, (Pt 4), 386-94. (48) Case, D. A.; Darden, T. A.; Gusarov, S.; Kovalenko, A.; Kollman, P. A., Amber 2015 (AmberTools15 and Amber14), University of California, San Francisco. 2015. (49) Hou, T.; Wang, J.; Li, Y.; Wang, W., Assessing the performance of the MM/PBSA and MM/GBSA methods. 1. The accuracy of binding free energy calculations based on molecular dynamics simulations. J. Chem. Inf. Model. 2011, 51, (1), 69-82. (50) Mysinger, M. M.; Carchia, M.; Irwin, J. J.; Shoichet, B. K., Directory of useful decoys, enhanced (DUD-E): better ligands and decoys for better benchmarking. J. Med. Chem. 2012, 55, (14), 6582-94. (51) Freitas, J.; Miller, N.; Mengeling, B. J.; Xia, M.; Huang, R.; Houck, K.; Rietjens, I. M.; Furlow, J. D.; Murk, A. J., Identification of thyroid hormone receptor active compounds using a quantitative high-throughput screening platform. Current chemical genomics and translational medicine 2014, 8, 36-46. (52) Browne, P.; Judson, R. S.; Casey, W. M.; Kleinstreuer, N. C.; Thomas, R. S., Screening Chemicals for Estrogen Receptor Bioactivity Using a Computational Model. Environ. Sci. Technol. 2015, 49, (14), 8804-14. (53) Huang, R.; Xia, M.; Nguyen, D.-T.; Zhao, T.; Sakamuru, S.; Zhao, J.; Shahane, S. A.; Rossoshek, A.; Simeonov, A., Tox21Challenge to build predictive models of nuclear receptor and stress response pathways as mediated by exposure to environmental chemicals and drugs. Frontiers in Environmental Science 2016, 3. (54) Tice, R. R.; Austin, C. P.; Kavlock, R. J.; Bucher, J. R., Improving the human hazard characterization of chemicals: a Tox21 update. Environ. Health Perspect. 2013, 121, (7), 756-65. (55) Schrödinger LigPrep, version 3.4, Schrödinger, LLC, New York, NY., 2015. (56) Schrödinger Glide, version 6.7, Schrödinger, LLC, New York, NY., 2015. (57) Jain, A. N.; Nicholls, A., Recommendations for evaluation of computational methods. J. Comput. Aided Mol. Des. 2008, 22, (3-4), 133-9. (58) Iakovleva, I.; Begum, A.; Pokrzywa, M.; Walfridsson, M.; Sauer-Eriksson, A. E.; Olofsson, A., The flavonoid luteolin, but not luteolin-7-O-glucoside, prevents a transthyretin mediated toxic response. PLoS One 2015, 10, (5), e0128222. (59) Hamers, T.; Kamstra, J. H.; Sonneveld, E.; Murk, A. J.; Kester, M. H.; Andersson, P. L.; Legler, J.; Brouwer, A., In vitro profiling of the endocrine-disrupting potency of brominated flame retardants. Toxicol. Sci. 2006, 92, (1), 157-73. (60) Trivella, D. B.; dos Reis, C. V.; Lima, L. M.; Foguel, D.; Polikarpov, I., Flavonoid interactions with human transthyretin: combined structural and thermodynamic analysis. J. Struct. Biol. 2012, 180, (1), 143-53.

ACS Paragon Plus Environment

21

Environmental Science & Technology

548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593

Page 22 of 30

(61) Yokoyama, T.; Kosaka, Y.; Mizuguchi, M., Inhibitory activities of propolis and its promising component, caffeic acid phenethyl ester, against amyloidogenesis of human transthyretin. J. Med. Chem. 2014, 57, (21), 8928-35. (62) Alhamadsheh, M. M.; Connelly, S.; Cho, A.; Reixach, N.; Powers, E. T.; Pan, D. W.; Wilson, I. A.; Kelly, J. W.; Graef, I. A., Potent kinetic stabilizers that prevent transthyretinmediated cardiomyocyte proteotoxicity. Sci. Transl. Med. 2011, 3, (97), 97ra81. (63) Wojtczak, A.; Cody, V.; Luft, J. R.; Pangborn, W., Structures of human transthyretin complexed with thyroxine at 2.0 A resolution and 3',5'-dinitro-N-acetyl-L-thyronine at 2.2 A resolution. Acta Crystallogr. D Biol. Crystallogr. 1996, 52, (Pt 4), 758-65. (64) Pospisil, P.; Ballmer, P.; Scapozza, L.; Folkers, G., Tautomerism in computer-aided drug design. J. Recept. Signal Transduct. 2003, 23, (4), 361-371. (65) Martin, Y. C., Let's not forget tautomers. J. Comput. Aided Mol. Des. 2009, 23, (10), 693704. (66) Shelley, J. C.; Cholleti, A.; Frye, L. L.; Greenwood, J. R.; Timlin, M. R.; Uchimaya, M., Epik: a software program for pKa prediction and protonation state generation for drug-like molecules. J. Comput. Aided Mol. Des. 2007, 21, (12), 681-91. (67) Friesner, R. A.; Banks, J. L.; Murphy, R. B.; Halgren, T. A.; Klicic, J. J.; Mainz, D. T.; Repasky, M. P.; Knoll, E. H.; Shelley, M.; Perry, J. K.; Shaw, D. E.; Francis, P.; Shenkin, P. S., Glide: A new approach for rapid, accurate docking and scoring. 1. Method and assessment of docking accuracy. J. Med. Chem. 2004, 47, (7), 1739-1749. (68) Muziol, T.; Cody, V.; Luft, J. R.; Pangborn, W.; Wojtczak, A., Complex of rat transthyretin with tetraiodothyroacetic acid refined at 2.1 and 1.8 A resolution. Acta Biochim. Pol. 2001, 48, (4), 877-84. (69) Ferguson, R. N.; Edelhoch, H.; Saroff, H. A.; Robbins, J.; Cahnmann, H. J., Negative cooperativity in the binding of thyroxine to human serum prealbumin. Preparation of tritiumlabeled 8-anilino-1-naphthalenesulfonic acid. Biochemistry 1975, 14, (2), 282-9. (70) Cheng, S. Y.; Pages, R. A.; Saroff, H. A.; Edelhoch, H.; Robbins, J., Analysis of thyroid hormone binding to human serum prealbumin by 8-anilinonaphthalene-1-sulfonate fluorescence. Biochemistry 1977, 16, (16), 3707-13. (71) Schneider, G., Virtual screening: an endless staircase? Nat. Rev. Drug Discov. 2010, 9, (4), 273-6. (72) Zhu, T.; Cao, S.; Su, P. C.; Patel, R.; Shah, D.; Chokshi, H. B.; Szukala, R.; Johnson, M. E.; Hevener, K. E., Hit identification and optimization in virtual screening: practical recommendations based on a critical literature analysis. J. Med. Chem. 2013, 56, (17), 6560-72. (73) Park, M. S.; Dessal, A. L.; Smrcka, A. V.; Stern, H. A., Evaluating docking methods for prediction of binding affinities of small molecules to the G protein betagamma subunits. J. Chem. Inf. Model. 2009, 49, (2), 437-43. (74) Warren, G. L.; Andrews, C. W.; Capelli, A. M.; Clarke, B.; LaLonde, J.; Lambert, M. H.; Lindvall, M.; Nevins, N.; Semus, S. F.; Senger, S.; Tedesco, G.; Wall, I. D.; Woolven, J. M.; Peishoff, C. E.; Head, M. S., A critical assessment of docking programs and scoring functions. J. Med. Chem. 2006, 49, (20), 5912-31. (75) Peterson, S. A.; Klabunde, T.; Lashuel, H. A.; Purkey, H.; Sacchettini, J. C.; Kelly, J. W., Inhibiting transthyretin conformational changes that lead to amyloid fibril formation. Proc. Natl. Acad. Sci. U. S. A. 1998, 95, (22), 12956-60. (76) Muziol, T.; Cody, V.; Wojtczak, A., Comparison of binding interactions of dibromoflavonoids with transthyretin. Acta Biochim. Pol. 2001, 48, (4), 885-92.

ACS Paragon Plus Environment

22

Page 23 of 30

594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639

Environmental Science & Technology

(77) Totrov, M.; Abagyan, R., Flexible ligand docking to multiple receptor conformations: a practical alternative. Curr. Opin. Struct. Biol. 2008, 18, (2), 178-84. (78) Frimurer, T. M.; Peters, G. H.; Iversen, L. F.; Andersen, H. S.; Moller, N. P.; Olsen, O. H., Ligand-induced conformational changes: improved predictions of ligand binding conformations and affinities. Biophys. J. 2003, 84, (4), 2273-81. (79) Bica, A.; Farinha, A.; Blume, H.; Barbosa, C. M., Determination of clonixin in plasma and urine by reversed-phase high-performance liquid chromatography. J. Chromatogr. A 2000, 889, (1-2), 135-41. (80) Marin-Water-Agency, Vegetation Management Plan 2010. www.marinwater.org/DocumentCenter/ (accessed Aug 6,2016). (81) Liao, C.; Liu, F.; Alomirah, H.; Loi, V. D.; Mohd, M. A.; Moon, H. B.; Nakata, H.; Kannan, K., Bisphenol S in urine from the United States and seven Asian countries: occurrence and human exposures. Environ. Sci. Technol. 2012, 46, (12), 6860-6. (82) Hall, J. C.; Deschamps, R. J. A.; Krieg, K. K., Immunoassays for the Detection of 2,4-D and Picloram in River Water and Urine. J. Agric. Food Chem. 1989, 37, (4), 981-984. (83) Rochester, J. R.; Bolden, A. L., Bisphenol S and F: A Systematic Review and Comparison of the Hormonal Activity of Bisphenol A Substitutes. Environ. Health Perspect. 2015, 123, (7), 643-50. (84) Kinch, C. D.; Ibhazehiebo, K.; Jeong, J. H.; Habibi, H. R.; Kurrasch, D. M., Low-dose exposure to bisphenol A and replacement bisphenol S induces precocious hypothalamic neurogenesis in embryonic zebrafish. Proc. Natl. Acad. Sci. U. S. A. 2015, 112, (5), 1475-80. (85) Mersha, M. D.; Patel, B. M.; Patel, D.; Richardson, B. N.; Dhillon, H. S., Effects of BPA and BPS exposure limited to early embryogenesis persist to impair non-associative learning in adults. Behav Brain Funct 2015, 11, 27. (86) Naderi, M.; Wong, M. Y.; Gholami, F., Developmental exposure of zebrafish (Danio rerio) to bisphenol-S impairs subsequent reproduction potential and hormonal balance in adults. Aquat. Toxicol. 2014, 148, 195-203. (87) Wang, W.; Abualnaja, K. O.; Asimakopoulos, A. G.; Covaci, A.; Gevao, B.; JohnsonRestrepo, B.; Kumosani, T. A.; Malarvannan, G.; Minh, T. B.; Moon, H. B.; Nakata, H.; Sinha, R. K.; Kannan, K., A comparative assessment of human exposure to tetrabromobisphenol A and eight bisphenols including bisphenol A via indoor dust ingestion in twelve countries. Environ. Int. 2015, 83, 183-91. (88) ATSDR Toxicological Profile for Dinitrocresols; USA Agency for Toxic Substances and Disease Registry (ATSDR), 1995. (89) Reddy, T. P.; Manczak, M.; Calkins, M. J.; Mao, P.; Reddy, A. P.; Shirendeb, U.; Park, B.; Reddy, P. H., Toxicity of neurons treated with herbicides and neuroprotection by mitochondriatargeted antioxidant SS31. Int. J. Environ. Res. Public Health 2011, 8, (1), 203-21. (90) Carney, E. W.; Billington, R.; Barlow, S. M., Developmental toxicity evaluation of triclopyr butoxyethyl ester and triclopyr triethylamine salt in the CD rat. Reprod. Toxicol. 2007, 23, (2), 165-74. (91) Schmidt, L.; Muller, J.; Goen, T., Simultaneous monitoring of seven phenolic metabolites of endocrine disrupting compounds (EDC) in human urine using gas chromatography with tandem mass spectrometry. Anal. Bioanal. Chem. 2013, 405, (6), 2019-29. (92) Fortenberry, G. Z.; Hu, H.; Turyk, M.; Barr, D. B.; Meeker, J. D., Association between urinary 3, 5, 6-trichloro-2-pyridinol, a metabolite of chlorpyrifos and chlorpyrifos-methyl, and serum T4 and TSH in NHANES 1999-2002. Sci. Total Environ. 2012, 424, 351-5.

ACS Paragon Plus Environment

23

Environmental Science & Technology

640 641 642 643 644 645 646 647 648 649 650 651 652 653 654

Page 24 of 30

(93) Gosselin, N. H.; Brunet, R. C.; Carrier, G.; Dosso, A., Worker exposures to triclopyr: risk assessment through measurements in urine samples. Ann. Occup. Hyg. 2005, 49, (5), 415-22. (94) Tran, A. T.; Hyne, R. V.; Doble, P., Determination of commonly used polar herbicides in agricultural drainage waters in Australia by HPLC. Chemosphere 2007, 67, (5), 944-53. (95) Botta, F.; Fauchon, N.; Blanchoud, H.; Chevreuil, M.; Guery, B., Phyt'Eaux Cites: application and validation of a programme to reduce surface water contamination with urban pesticides. Chemosphere 2012, 86, (2), 166-76. (96) Battaglin, W. A.; Rice, K. C.; Focazio, M. J.; Salmons, S.; Barry, R. X., The occurrence of glyphosate, atrazine, and other pesticides in vernal pools and adjacent streams in Washington, DC, Maryland, Iowa, and Wyoming, 2005-2006. Environ. Monit. Assess. 2009, 155, (1-4), 281307. (97) Tagert, M. L.; Massey, J. H.; Shaw, D. R., Water quality survey of Mississippi's Upper Pearl River. Sci. Total Environ. 2014, 481, 564-73. (98) Purkey, H. E.; Dorrell, M. I.; Kelly, J. W., Evaluating the binding selectivity of transthyretin amyloid fibril inhibitors in blood plasma. Proc. Natl. Acad. Sci. U. S. A. 2001, 98, (10), 5566-71.

655 656 657 658

ACS Paragon Plus Environment

24

Page 25 of 30

659 660 661

Environmental Science & Technology

Graphical abstract

662 663 664

ACS Paragon Plus Environment

25

Environmental Science & Technology

Page 26 of 30

665 666

Table 1. Studied compounds with abbreviations, chemical abstract services (CAS) registry

667

numbers, usage, and binding affinities (Kd) determined using isothermal titration calorimetry. Compounds

Abbreviations

CAS

Usage

Kd (µM)a

3,3',5,5'-tetraiodo-L-thyronine

T4

51-48-9

Positive Control

0.09b

2-nitro-5-(2-chloro-4Acifluorfen trifluoromethylphenoxy)benzoic acid

50594-66-6

Herbicide

ND

2,2'-dihydroxy-4,4'dimethoxybenzophenone

BP6

131-54-4

UV absorber

ND

4,4'-dihydroxydiphenyl sulfone

BPS

80-09-1

Plasticizer

52

2-(3-chloro-2methylanilino)pyridine-3carboxylic acid

Clonixin

17737-65-4

Pharmaceutic 0.26 al

2,6-dinitro-p-cresol

DNPC

609-93-8

Polymerizati on inhibitor

1.3

diphenolic acid

DPA

126-00-1

Plasticizer

ND

2-[(4-amino-3,5-dichloro-6fluoro-2-pyridinyl)oxy]acetic acid

Fluroxypyr

69377-81-7

Herbicide

45

L-γ-glutamyl-p-nitroanilide

GPNA

67953-08-6

Food additive

ND

2-[4-(methylsulfonyl)-2nitrobenzoyl]-1,3cyclohexanedione

Mesotrione

104206-82-8

Herbicide

99

3,5,6-trichloro-4-aminopicolinic Picloram acid

1918-02-1

Herbicide

63

1-(4-sulfophenyl)-3-carboxy-5pyrazolone

PyT

118-47-8

Dye

ND

3,5,6-trichloro-2pyridinyloxyacetic acid

Triclopyr

55335-06-3

Herbicide

4.6

ACS Paragon Plus Environment

26

Page 27 of 30

668 669 670

Environmental Science & Technology

a

ND refers to non-detected affinity. bThe binding affinity of T4 is only an approximate value due to its thermal instability and poor solubility in the isothermal titration calorimetry buffer. The approximated binding affinity is similar to the value of 84 nM reported previously.98

671 672

ACS Paragon Plus Environment

27

Environmental Science & Technology

Page 28 of 30

673

674 675

Figure 1. (A) The TTR monomers in the dimer structure are shown as ribbons and are labeled A

676

and B. The symmetry-related monomers are labeled A´ and B´. Locations of the thyroxine

677

binding sites (TBSs) are shown as gray surfaces. (B) Close-up view of the crystallographic

678

binding conformations of (a) BP2, (b) PFOA, and (c) PFOS in the TBS. The refined (2|Fo|−|Fc|)

679

electron density is shown in gray mesh at 1σ level, and the anomalous difference map showing

680

the position of the sulfur atoms in PFOS is shown in violet mesh at 3σ level. The water

681

molecules present around the ligands are shown as red spheres.

682

ACS Paragon Plus Environment

28

Page 29 of 30

Environmental Science & Technology

683

684 685

Figure 2. Structures of active compounds and thermograms of their binding to TTR as

686

determined by isothermal titration calorimetry. The abbreviations of the compounds are given in

687

Table 1.

688

ACS Paragon Plus Environment

29

Environmental Science & Technology

Page 30 of 30

689

690 691

Figure 3. Crystallographic binding conformations of the selected active compounds and T4 in

692

the thyroid-binding site of TTR; (a) clonixin, (b) DNPC (in the AA’ interface), (c) DNPC (in the

693

BB’ interface), (d) triclopyr, (e) T4 (PDB ID: 2ROX, for comparison), and (f) BPS. The refined

694

(2|Fo|−|Fc|) electron density is shown in gray mesh at 1σ level. The water molecules present

695

around the ligands are shown as red spheres. The electron density of the second ring of BPS was

696

poorly defined probably due to high mobility.

697

ACS Paragon Plus Environment

30